J. E. Chang and S. J. Turley, Stromal infrastructure of the lymph node and coordination of immunity, Trends Immunol, vol.36, pp.30-39, 2015.

C. Mionnet, I. Mondor, A. Jorquera, M. Loosveld, J. Maurizio et al., Identification of a new stromal cell type involved in the regulation of inflamed B cell follicles, PLoS Biol, vol.11, p.1001672, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-02131392

H. Huang, A. Rivas-caicedo, F. Renevey, H. Cannelle, E. Peranzoni et al., Identification of a new subset of lymph node stromal cells involved in regulating plasma cell homeostasis, Proc. Natl. Acad. Sci. U.S.a, vol.115, pp.6826-6835, 2018.

D. Malhotra, A. L. Fletcher, J. Astarita, V. Lukacs-kornek, P. Tayalia et al., Transcriptional profiling of stroma from inflamed and resting lymph nodes defines immunological hallmarks, Nat. Immunol, vol.13, pp.499-510, 2012.

L. B. Rodda, E. Lu, M. L. Bennett, C. L. Sokol, X. Wang et al., Single-Cell RNA Sequencing of Lymph Node Stromal Cells Reveals NicheAssociated Heterogeneity, Immunity, vol.48, pp.1014-1028, 2018.

C. Bénézech, E. Mader, G. Desanti, M. Khan, K. Nakamura et al., Lymphotoxin-? receptor signaling through NF-?B2-RelB pathway reprograms adipocyte precursors as lymph node stromal cells, Immunity, vol.37, pp.721-734, 2012.

M. Jarjour, A. Jorquera, I. Mondor, S. Wienert, P. Narang et al., Fate mapping reveals origin and dynamics of lymph node follicular dendritic cells, J. Exp. Med, vol.211, pp.1109-1122, 2014.

K. M. Sitnik, K. Wendland, H. Weishaupt, H. Uronen-hansson, A. J. White et al., Context-Dependent Development of Lymphoid Stroma from Adult CD34(+) Adventitial Progenitors, Cell Rep, vol.14, pp.2375-2388, 2016.

N. J. Krautler, V. Kana, J. Kranich, Y. Tian, D. Perera et al., Follicular dendritic cells emerge from ubiquitous perivascular precursors, Cell, vol.150, pp.194-206, 2012.

Q. Chai, L. Onder, E. Scandella, C. Gil-cruz, C. Perez-shibayama et al., Maturation of lymph node fibroblastic reticular cells from myofibroblastic precursors is critical for antiviral immunity, Immunity, vol.38, pp.1013-1024, 2013.

R. Golub, J. Tan, T. Watanabe, and A. Brendolan, Origin and Immunological Functions of Spleen Stromal Cells, Trends Immunol, vol.39, pp.503-514, 2018.
URL : https://hal.archives-ouvertes.fr/pasteur-01768627

X. Wang, B. Cho, K. Suzuki, Y. Xu, J. A. Green et al., Follicular dendritic cells help establish follicle identity and promote B cell retention in germinal centers, J. Exp. Med, vol.208, pp.2497-2510, 2011.

R. Endres, M. B. Alimzhanov, T. Plitz, A. Fütterer, M. H. Kosco-vilbois et al., Mature follicular dendritic cell networks depend on expression of lymphotoxin beta receptor by radioresistant stromal cells and of lymphotoxin beta and tumor necrosis factor by B cells, J. Exp. Med, vol.189, pp.159-168, 1999.

K. M. Ansel, V. N. Ngo, P. L. Hyman, S. A. Luther, R. Förster et al., A chemokine-driven positive feedback loop organizes lymphoid follicles, Nature, vol.406, pp.309-314, 2000.

B. A. Heesters, R. C. Myers, and M. C. Carroll, Follicular dendritic cells: dynamic antigen libraries, Nat. Rev. Immunol, vol.14, pp.495-504, 2014.

C. R. Nowosad, K. M. Spillane, and P. Tolar, Germinal center B cells recognize antigen through a specialized immune synapse architecture, Nat. Immunol, vol.17, pp.870-877, 2016.

L. Mesin, J. Ersching, and G. D. Victora, Germinal Center B Cell Dynamics, vol.45, pp.471-482, 2016.

G. Koopman, R. M. Keehnen, E. Lindhout, W. Newman, Y. Shimizu et al., Adhesion through the LFA-1 (CD11a/CD18)-ICAM-1 (CD54) and the VLA-4 (CD49d)-VCAM-1 (CD106) pathways prevents apoptosis of germinal center B cells, J. Immunol, vol.152, pp.3760-3767, 1994.

S. Yoon, X. Zhang, P. Berner, B. Blom, and Y. S. Choi, Notch ligands expressed by follicular dendritic cells protect germinal center B cells from apoptosis, J. Immunol, vol.183, pp.352-358, 2009.

J. Kim, D. W. Kim, W. Chang, J. Choe, J. Kim et al., Wnt5a is secreted by follicular dendritic cells to protect germinal center B cells via Wnt/Ca2+/NFAT/NF-?B-B cell lymphoma 6 signaling, J. Immunol, vol.188, pp.182-189, 2012.

R. Sacedón, B. Díez, V. Nuñez, C. Hernández-lópez, C. Gutierrez-frías et al., Sonic hedgehog is produced by follicular dendritic cells and protects germinal center B cells from apoptosis, J. Immunol, vol.174, pp.1456-1461, 2005.

C. Park, S. Yoon, R. J. Armitage, and Y. S. Choi, Follicular dendritic cells produce IL-15 that enhances germinal center B cell proliferation in membranebound form, J. Immunol, vol.173, pp.6676-6683, 2004.

L. Gorelik, K. Gilbride, M. Dobles, S. L. Kalled, D. Zandman et al., Normal B cell homeostasis requires B cell activation factor production by radiationresistant cells, J. Exp. Med, vol.198, pp.937-945, 2003.

K. Suzuki, M. Maruya, S. Kawamoto, K. Sitnik, H. Kitamura et al., The sensing of environmental stimuli by follicular dendritic cells promotes immunoglobulin A generation in the gut, Immunity, vol.33, pp.71-83, 2010.

J. Kranich, N. J. Krautler, E. Heinen, M. Polymenidou, C. Bridel et al., Follicular dendritic cells control engulfment of apoptotic bodies by secreting Mfge8, J. Exp. Med, vol.205, pp.1293-1302, 2008.

L. B. Rodda, O. Bannard, B. Ludewig, T. Nagasawa, and J. G. Cyster, Phenotypic and Morphological Properties of Germinal Center Dark Zone Cxcl12-Expressing Reticular Cells, J. Immunol, vol.195, pp.4781-4791, 2015.

O. Bannard, R. M. Horton, C. D. Allen, J. An, T. Nagasawa et al., Germinal center centroblasts transition to a centrocyte phenotype according to a timed program and depend on the dark zone for effective selection, Immunity, vol.39, pp.912-924, 2013.

D. Suan, C. Sundling, and R. Brink, Plasma cell and memory B cell differentiation from the germinal center, Curr. Opin. Immunol, vol.45, pp.97-102, 2017.

E. P. Tjin, R. J. Bende, P. W. Derksen, A. Van-huijstee, H. Kataoka et al.,

. Spaargaren, Follicular dendritic cells catalyze hepatocyte growth factor (HGF) activation in the germinal center microenvironment by secreting the serine protease HGF activator, J. Immunol, vol.175, pp.2807-2813, 2005.

T. Katakai, Marginal reticular cells: a stromal subset directly descended from the lymphoid tissue organizer, Front Immunol, vol.3, p.200, 2012.

R. Roozendaal, T. R. Mempel, L. A. Pitcher, S. F. Gonzalez, A. Verschoor et al., Conduits mediate transport of low-molecular-weight antigen to lymph node follicles, Immunity, vol.30, pp.264-276, 2009.

K. Sato, S. Honda, A. Shibuya, and K. Shibuya, Cutting Edge: Identification of Marginal Reticular Cells as Phagocytes of Apoptotic B Cells in Germinal Centers, J. Immunol, vol.200, pp.3691-3696, 2018.

V. Cremasco, M. C. Woodruff, L. Onder, J. Cupovic, J. M. Nieves-bonilla et al., B cell homeostasis and follicle confines are governed by fibroblastic reticular cells, Nat. Immunol, vol.15, pp.973-981, 2014.

Y. O. Alexandre and S. N. Mueller, Stromal cell networks coordinate immune response generation and maintenance, Immunol. Rev, vol.283, pp.77-85, 2018.

M. Bajénoff, J. G. Egen, L. Y. Koo, J. P. Laugier, F. Brau et al., Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes, Immunity, vol.25, pp.989-1001, 2006.

Y. Zhang, L. Tech, L. A. George, A. Acs, R. E. Durrett et al., Plasma cell output from germinal centers is regulated by signals from Tfh and stromal cells, J. Exp. Med, vol.215, pp.1227-1243, 2018.

J. L. Astarita, V. Cremasco, J. Fu, M. C. Darnell, J. R. Peck et al., The CLEC-2-podoplanin axis controls the contractility of fibroblastic reticular cells and lymph node microarchitecture, Nat. Immunol, vol.16, pp.75-84, 2015.

L. K. Dubey, L. Lebon, I. Mosconi, C. Yang, E. Scandella et al., Lymphotoxin-Dependent B Cell-FRC Crosstalk Promotes De Novo Follicle Formation and Antibody Production following Intestinal Helminth Infection, Cell Rep, vol.15, pp.1527-1541, 2016.

J. L. Gregory, A. Walter, Y. O. Alexandre, J. L. Hor, R. Liu et al., Infection Programs Sustained Lymphoid Stromal Cell Responses and Shapes Lymph Node Remodeling upon Secondary Challenge, Cell Rep, vol.18, pp.406-418, 2017.

F. D. Brown and S. J. Turley, Fibroblastic reticular cells: organization and regulation of the T lymphocyte life cycle, J. Immunol, vol.194, pp.1389-1394, 2015.

V. Lukacs-kornek, D. Malhotra, A. L. Fletcher, S. E. Acton, K. G. Elpek et al., Regulated release of nitric oxide by nonhematopoietic stroma controls expansion of the activated T cell pool in lymph nodes, Nat. Immunol, vol.12, pp.1096-1104, 2011.

K. Knoblich, S. C. Migoni, S. M. Siew, E. Jinks, B. Kaul et al., The human lymph node microenvironment unilaterally regulates T-cell activation and differentiation, PLoS Biol, vol.16, p.2005046, 2018.

H. Maby-el-hajjami, P. Amé-thomas, C. Pangault, O. Tribut, J. Devos et al., Functional alteration of the lymphoma stromal cell niche by the cytokine context: role of indoleamine-2,3 dioxygenase, Cancer Res, vol.69, pp.3228-3237, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00869384

L. Verdière, F. Mourcin, and K. Tarte, Microenvironment signaling driving lymphomagenesis, Curr. Opin. Hematol, vol.25, pp.335-345, 2018.

P. Amé-thomas and K. Tarte, The yin and the yang of follicular lymphoma cell niches: role of microenvironment heterogeneity and plasticity, Semin. Cancer Biol, vol.24, pp.23-32, 2014.

F. Vega, L. J. Medeiros, W. Lang, A. Mansoor, C. Bueso-ramos et al., The stromal composition of malignant lymphoid aggregates in bone marrow: variations in architecture and phenotype in different B-cell tumours, Br. J. Haematol, vol.117, pp.569-576, 2002.

S. Pandey, F. Mourcin, T. Marchand, S. Nayar, M. Guirriec et al., IL-4/CXCL12 loop is a key regulator of lymphoid stroma function in follicular lymphoma, Blood, vol.129, pp.2507-2518, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01555850

S. Sangaletti, C. Tripodo, P. Portararo, M. Dugo, C. Vitali et al., Stromal niche communalities underscore the contribution of the matricellular protein SPARC to B-cell development and lymphoid malignancies, Oncoimmunology, vol.3, p.28989, 2014.

P. Amé-thomas, H. Maby-el-hajjami, C. Monvoisin, R. Jean, D. Monnier et al., Human mesenchymal stem cells isolated from bone marrow and lymphoid organs support tumor B-cell growth: role of stromal cells in follicular lymphoma pathogenesis, Blood, vol.109, pp.693-702, 2007.

M. Mraz, C. S. Zent, A. K. Church, D. F. Jelinek, X. Wu et al., Bone marrow stromal cells protect lymphoma B-cells from rituximab-induced apoptosis and targeting integrin ?-4-?-1 (VLA-4) with natalizumab can overcome this resistance, Br. J. Haematol, vol.155, pp.53-64, 2011.

J. A. Green and J. G. Cyster, S1PR2 links germinal center confinement and growth regulation, vol.247, pp.36-51, 2012.

A. Bouska, W. Zhang, Q. Gong, J. Iqbal, A. Scuto et al., Combined copy number and mutation analysis identifies oncogenic pathways associated with transformation of follicular lymphoma, Leukemia, vol.31, pp.83-91, 2017.

F. Mourcin, C. Pangault, R. Amin-ali, P. Amé-thomas, and K. Tarte, Stromal cell contribution to human follicular lymphoma pathogenesis, Front Immunol, vol.3, p.280, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00869146

M. Weissmann, G. Arvatz, N. Horowitz, S. Feld, I. Naroditsky et al., Heparanase-neutralizing antibodies attenuate lymphoma tumor growth and metastasis, Proc. Natl. Acad. Sci. U.S.A, vol.113, pp.704-709, 2016.

F. Guilloton, G. Caron, C. Ménard, C. Pangault, P. Amé-thomas et al., Mesenchymal stromal cells orchestrate follicular lymphoma cell niche through the CCL2-dependent recruitment and polarization of monocytes, Blood, vol.119, pp.2556-2567, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00665887

G. Epron, P. Amé-thomas, J. L. Priol, C. Pangault, J. Dulong et al., Monocytes and T cells cooperate to favor normal and follicular lymphoma Bcell growth: role of IL-15 and CD40L signaling, Leukemia, vol.26, pp.139-148, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00869028

R. Amin, F. Mourcin, F. Uhel, C. Pangault, P. Ruminy et al., DC-SIGN-expressing macrophages trigger activation of mannosylated IgM B-cell receptor in follicular lymphoma, Blood, vol.126, pp.1911-1920, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01187301

M. Grégoire, F. Guilloton, C. Pangault, F. Mourcin, P. Sok et al., Neutrophils trigger a NF-?B dependent polarization of tumor-supportive stromal cells in germinal center B-cell lymphomas, Oncotarget, vol.6, pp.16471-16487, 2015.

M. Travert, P. Amé-thomas, C. Pangault, A. Morizot, O. Micheau et al., CD40 ligand protects from TRAIL-induced apoptosis in follicular lymphomas through NF-kappaB activation and up-regulation of c-FLIP and BclxL, J. Immunol, vol.181, pp.1001-1011, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00869465

M. J. Lee, S. Y. Park, J. H. Ko, H. J. Lee, J. S. Ryu et al., Mesenchymal stromal cells promote B-cell lymphoma in lacrimal glands by inducing immunosuppressive microenvironment, Oncotarget, vol.8, pp.66281-66292, 2017.

K. Chang, X. Huang, L. J. Medeiros, and D. Jones, Germinal centre-like versus undifferentiated stromal immunophenotypes in follicular lymphoma, J. Pathol, vol.201, pp.404-412, 2003.

G. Pepe, A. D. Napoli, C. Cippitelli, S. Scarpino, E. Pilozzi et al., Reduced lymphotoxin-beta production by tumour cells is associated with loss of follicular dendritic cell phenotype and diffuse growth in follicular lymphoma, J Pathol Clin Res, vol.4, pp.124-134, 2018.

A. Costa, Y. Kieffer, A. Scholer-dahirel, F. Pelon, B. Bourachot et al., Fibroblast Heterogeneity and Immunosuppressive Environment in Human Breast Cancer, Cancer Cell, vol.33, pp.463-479, 2018.

M. Boice, D. Salloum, F. Mourcin, V. Sanghvi, R. Amin et al., Loss of the HVEM Tumor Suppressor in Lymphoma and Restoration by Modified CAR-T Cells, vol.167, pp.405-418, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01405853

F. Fiore, M. S. Von-bergwelt-baildon, U. Drebber, M. Beyer, A. Popov et al., Dendritic cells are significantly reduced in non-Hodgkin's lymphoma and express less CCR7 and CD62L, Leuk. Lymphoma, vol.47, pp.613-622, 2006.

P. Gravelle, C. Jean, J. Familiades, E. Decaup, A. Blanc et al., Cell growth in aggregates determines gene expression, proliferation, survival, chemoresistance, and sensitivity to immune effectors in follicular lymphoma, Am. J. Pathol, vol.184, pp.282-295, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-00905913

Y. F. Tian, H. Ahn, R. S. Schneider, S. N. Yang, L. Roman-gonzalez et al., Integrin-specific hydrogels as adaptable tumor organoids for malignant B and T cells, Biomaterials, vol.73, pp.110-119, 2015.

F. Apoorva, A. M. Loiben, S. B. Shah, A. Purwada, L. Fontan et al., How Biophysical Forces Regulate Human B Cell Lymphomas, Cell Rep, vol.23, pp.499-511, 2018.

R. G. Mannino, A. N. Santiago-miranda, P. Pradhan, Y. Qiu, J. C. Mejias et al., 3D microvascular model recapitulates the diffuse large B-cell lymphoma tumor microenvironment in vitro, Lab Chip, vol.17, pp.407-414, 2017.

P. Milpied, B. Nadel, and S. Roulland, Premalignant cell dynamics in indolent B-cell malignancies, Curr. Opin. Hematol, vol.22, pp.388-396, 2015.

A. Egle, A. W. Harris, M. L. Bath, L. O'reilly, and S. Cory, VavP-Bcl2 transgenic mice develop follicular lymphoma preceded by germinal center hyperplasia, Blood, vol.103, pp.2276-2283, 2004.

Y. Jiang, A. Ortega-molina, H. Geng, H. Ying, K. Hatzi et al., CREBBP Inactivation Promotes the Development of HDAC3-Dependent Lymphomas, Cancer Discov, vol.7, pp.38-53, 2017.

J. Zhang, D. Dominguez-sola, S. Hussein, J. Lee, A. B. Holmes et al., Disruption of KMT2D perturbs germinal center B cell development and promotes lymphomagenesis, Nat. Med, vol.21, pp.1190-1198, 2015.

S. H. Swerdlow, E. Campo, S. A. Pileri, N. L. Harris, H. Stein et al., The 2016 revision of the World Health Organization classification of lymphoid neoplasms, Blood, vol.127, pp.2375-2390, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01800015

K. Takata, Y. Sato, N. Nakamura, Y. Y. Kikuti, K. Ichimura et al., Duodenal and nodal follicular lymphomas are distinct: the former lacks activation-induced cytidine deaminase and follicular dendritic cells despite ongoing somatic hypermutations, Mod. Pathol, vol.22, pp.940-949, 2009.

S. Araf, J. Wang, K. Korfi, C. Pangault, E. Kotsiou et al., Genomic profiling reveals spatial intra-tumor heterogeneity in follicular lymphoma, Leukemia, vol.32, pp.1258-1263, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01815695

H. Rajnai, C. Bödör, Z. Balogh, E. Gagyi, J. Csomor et al., Impact of the reactive microenvironment on the bone marrow involvement of follicular lymphoma, Histopathology, vol.60, pp.66-75, 2012.

A. Bognár, B. Csernus, C. Bödör, L. Reiniger, A. Szepesi et al., Clonal selection in the bone marrow involvement of follicular lymphoma, Leukemia, vol.19, pp.1656-1662, 2005.

M. Wartenberg, P. Vasil, C. M. Bueschenfelde, G. Ott, A. Rosenwald et al., Somatic hypermutation analysis in follicular lymphoma provides evidence suggesting bidirectional cell migration between lymph node and bone marrow during disease progression and relapse, Haematologica, vol.98, pp.1433-1441, 2013.

S. Mariathasan, S. J. Turley, D. Nickles, A. Castiglioni, K. Yuen et al., TGF? attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells, Nature, vol.554, pp.544-548, 2018.